Correlation Engine 2.0
Clear Search sequence regions


  • alloantigen (1)
  • allograft (7)
  • antibodies (3)
  • CD40 (2)
  • cd40l (3)
  • cell (1)
  • female (1)
  • impaired (3)
  • intracellular (1)
  • islets langerhans (1)
  • JNK (4)
  • ligand (1)
  • lymphocyte (1)
  • male (1)
  • mice (5)
  • mice balb c (1)
  • mice knockout (1)
  • mitogen (2)
  • phase (1)
  • protein kinases (2)
  • t cell receptor (1)
  • t lymphocytes (12)
  • TNF (2)
  • tnf receptor (1)
  • TRAF2 (11)
  • vitro (1)
  • Sizes of these terms reflect their relevance to your search.

    Administration of anti-CD40 ligand (CD40L) antibodies has been reported to allow long-term islet allograft survival in non-human primates without the need for exogenous immunosuppression. However, the use of anti-CD40L antibodies was associated with thromboembolic complications. Targeting downstream intracellular components shared between CD40 and other TNF family co-stimulatory molecules could bypass these complications. TNF receptor associated factor 2 (TRAF2) integrates multiple TNF receptor family signalling pathways that are critical for T cell activation and may be a central node of alloimmune responses. T cell-specific Traf2-deficient mice (Traf2TKO) were generated to define the role of TRAF2 in CD4+ T cell effector responses that mediate islet allograft rejection in vivo. In vitro allograft responses were tested using mixed lymphocyte reactions and analysis of IFN-γ and granzyme B effector molecule expression. T cell function was assessed using anti-CD3/CD28-mediated proliferation and T cell polarisation studies. Traf2TKO mice exhibited permanent survival of full MHC-mismatched pancreatic islet allografts without exogenous immunosuppression. Traf2TKO CD4+ T cells exhibited reduced proliferation, activation and acquisition of effector function following T cell receptor stimulation; however, both Traf2TKO CD4+ and CD8+ T cells exhibited impaired alloantigen-mediated proliferation and acquisition of effector function. In polarisation studies, Traf2TKO CD4+ T cells preferentially converted to a T helper (Th)2 phenotype, but exhibited impaired Th17 differentiation. Without TRAF2, thymocytes exhibited dysregulated TNF-mediated induction of c-Jun N-terminal kinase (JNK) and canonical NFκB pathways. Critically, targeting TRAF2 in T cells did not impair the acute phase of CD8-dependent viral immunity. These data highlight a specific requirement for a TRAF2-NFκB and TRAF2-JNK signalling cascade in T cell activation and effector function in rejecting islet allografts. Targeting TRAF2 may be useful as a therapeutic approach for immunosuppression-free islet allograft survival that avoids the thromboembolic complications associated with the use of anti-CD40L antibodies.

    Citation

    Jeanette E Villanueva, Stacey N Walters, Mitsuru Saito, Elisabeth K Malle, Nathan W Zammit, Katherine A Watson, Robert Brink, Nicole L La Gruta, Stephen I Alexander, Shane T Grey. Targeted deletion of Traf2 allows immunosuppression-free islet allograft survival in mice. Diabetologia. 2017 Apr;60(4):679-689

    Expand section icon Mesh Tags

    Expand section icon Substances


    PMID: 28062921

    View Full Text